Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Cell ; 146(2): 318-31, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21757228

RESUMO

Patient-specific induced pluripotent stem cells (iPSCs) derived from somatic cells provide a unique tool for the study of human disease, as well as a promising source for cell replacement therapies. One crucial limitation has been the inability to perform experiments under genetically defined conditions. This is particularly relevant for late age onset disorders in which in vitro phenotypes are predicted to be subtle and susceptible to significant effects of genetic background variations. By combining zinc finger nuclease (ZFN)-mediated genome editing and iPSC technology, we provide a generally applicable solution to this problem, generating sets of isogenic disease and control human pluripotent stem cells that differ exclusively at either of two susceptibility variants for Parkinson's disease by modifying the underlying point mutations in the α-synuclein gene. The robust capability to genetically correct disease-causing point mutations in patient-derived hiPSCs represents significant progress for basic biomedical research and an advance toward hiPSC-based cell replacement therapies.


Assuntos
Doença de Parkinson/patologia , Células-Tronco Pluripotentes , Mutação Puntual , Linhagem Celular , Células-Tronco Embrionárias , Engenharia Genética , Estudo de Associação Genômica Ampla , Humanos , Mutagênese , Oligonucleotídeos/metabolismo , alfa-Sinucleína/genética
2.
J Biol Chem ; 293(29): 11341-11357, 2018 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-29858247

RESUMO

Mounting evidence suggests that alterations in cholesterol homeostasis are involved in Alzheimer's disease (AD) pathogenesis. Amyloid precursor protein (APP) or multiple fragments generated by proteolytic processing of APP have previously been implicated in the regulation of cholesterol metabolism. However, the physiological function of APP in regulating lipoprotein homeostasis in astrocytes, which are responsible for de novo cholesterol biosynthesis and regulation in the brain, remains unclear. To address this, here we used CRISPR/Cas9 genome editing to generate isogenic APP-knockout (KO) human induced pluripotent stem cells (hiPSCs) and differentiated them into human astrocytes. We found that APP-KO astrocytes have reduced cholesterol and elevated levels of sterol regulatory element-binding protein (SREBP) target gene transcripts and proteins, which were both downstream consequences of reduced lipoprotein endocytosis. To elucidate which APP fragments regulate cholesterol homeostasis and to examine whether familial AD mutations in APP affect lipoprotein metabolism, we analyzed an isogenic allelic series harboring the APP Swedish and APP V717F variants. Only astrocytes homozygous for the APP Swedish (APPSwe/Swe) mutation, which had reduced full-length APP (FL APP) due to increased ß-secretase cleavage, recapitulated the APP-KO phenotypes. Astrocytic internalization of ß-amyloid (Aß), another ligand for low-density lipoprotein (LDL) receptors, was also impaired in APP-KO and APPSwe/Swe astrocytes. Finally, impairing cleavage of FL APP through ß-secretase inhibition in APPSwe/Swe astrocytes reversed the LDL and Aß endocytosis defects. In conclusion, FL APP is involved in the endocytosis of LDL receptor ligands and is required for proper cholesterol homeostasis and Aß clearance in human astrocytes.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Astrócitos/metabolismo , Colesterol/metabolismo , Lipoproteínas/metabolismo , Precursor de Proteína beta-Amiloide/genética , Astrócitos/citologia , Sistemas CRISPR-Cas , Linhagem Celular , Endocitose , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Receptores de LDL/metabolismo , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo
3.
Future Sci OA ; 10(1): FSO964, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38817352

RESUMO

Aim: We explored the generation of human induced pluripotent stem cells (iPSCs) solely through the transcriptional activation of endogenous genes by CRISPR activation (CRISPRa). Methods: Minimal number of human-specific guide RNAs targeting a limited set of loci were used with a unique cocktail of small molecules (CRISPRa-SM). Results: iPSC clones were efficiently generated by CRISPRa-SM, expressed general and naive iPSC markers and clustered with high-quality iPSCs generated using conventional reprogramming methods. iPSCs showed genomic stability and robust pluripotent potential as assessed by in vitro and in vivo. Conclusion: CRISPRa-SM-generated human iPSCs by direct and multiplexed loci activation facilitating a unique and potentially safer cellular reprogramming process to aid potential applications in cellular therapy and regenerative medicine.


Combined chemical and CRISPRa-mediated approach leads to efficient generation of human iPSCs.

4.
J Neurosci ; 30(49): 16469-74, 2010 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-21147986

RESUMO

Loss of dopaminergic neurons is primarily responsible for the onset and progression of Parkinson's disease (PD); thus, neuroprotective and/or neuroregenerative strategies remain critical to the treatment of this increasingly prevalent disease. Here we explore a novel approach to neurotrophic factor-based therapy by engineering zinc finger protein transcription factors (ZFP TFs) that activate the expression of the endogenous glial cell line-derived neurotrophic factor (GDNF) gene. We show that GDNF activation can be achieved with exquisite genome-wide specificity. Furthermore, in a rat model of PD, striatal delivery of an adeno-associated viral vector serotype 2 encoding the GDNF activator resulted in improvements in forelimb akinesia, sensorimotor neglect, and amphetamine-induced rotations caused by 6-hydroxydopamine (6-OHDA) lesion. Our results suggest that an engineered ZFP TF can drive sufficient GDNF expression in the brain to provide functional neuroprotection against 6-OHDA; therefore, targeted activation of the endogenous gene may provide a method for delivering appropriate levels of GDNF to PD patients.


Assuntos
Terapia Genética/métodos , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Doença de Parkinson/terapia , Engenharia de Proteínas/métodos , Anfetamina/administração & dosagem , Animais , Linhagem Celular , Modelos Animais de Doenças , Dopaminérgicos/administração & dosagem , Ensaio de Imunoadsorção Enzimática/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Vetores Genéticos/fisiologia , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/biossíntese , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/genética , Proteínas de Fluorescência Verde/genética , Haplorrinos , Humanos , Lentivirus/fisiologia , Camundongos , Análise em Microsséries/métodos , Atividade Motora/efeitos dos fármacos , Oxidopamina/toxicidade , Doença de Parkinson/complicações , Doença de Parkinson/etiologia , RNA Mensageiro/metabolismo , Ratos , Fatores de Tempo , Transfecção , Tirosina 3-Mono-Oxigenase/metabolismo , Dedos de Zinco/genética
5.
Nat Genet ; 51(12): 1691-1701, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31740836

RESUMO

In the mammalian genome, the clustered protocadherin (cPCDH) locus provides a paradigm for stochastic gene expression with the potential to generate a unique cPCDH combination in every neuron. Here we report a chromatin-based mechanism that emerges during the transition from the naive to the primed states of cell pluripotency and reduces, by orders of magnitude, the combinatorial potential in the human cPCDH locus. This mechanism selectively increases the frequency of stochastic selection of a small subset of cPCDH genes after neuronal differentiation in monolayers, 10-month-old cortical organoids and engrafted cells in the spinal cords of rats. Signs of these frequent selections can be observed in the brain throughout fetal development and disappear after birth, except in conditions of delayed maturation such as Down's syndrome. We therefore propose that a pattern of limited cPCDH-gene expression diversity is maintained while human neurons still retain fetal-like levels of maturation.


Assuntos
Caderinas/genética , Cromatina/genética , Síndrome de Down/patologia , Células-Tronco Pluripotentes Induzidas/citologia , Neurônios/fisiologia , Adulto , Animais , Astrócitos/citologia , Astrócitos/fisiologia , Encéfalo/citologia , Encéfalo/embriologia , Diferenciação Celular , Linhagem Celular , Síndrome de Down/genética , Regulação da Expressão Gênica , Histonas/genética , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Células-Tronco Pluripotentes Induzidas/transplante , Camundongos , Pessoa de Meia-Idade , Neurônios/citologia , Regiões Promotoras Genéticas , Ratos , Análise de Célula Única , Medula Espinal/citologia , Medula Espinal/transplante , Transplante Heterólogo
6.
Cell Stem Cell ; 24(3): 363-375.e9, 2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30686764

RESUMO

Genetic, epidemiologic, and biochemical evidence suggests that predisposition to Alzheimer's disease (AD) may arise from altered cholesterol metabolism, although the molecular pathways that may link cholesterol to AD phenotypes are only partially understood. Here, we perform a phenotypic screen for pTau accumulation in AD-patient iPSC-derived neurons and identify cholesteryl esters (CE), the storage product of excess cholesterol, as upstream regulators of Tau early during AD development. Using isogenic induced pluripotent stem cell (iPSC) lines carrying mutations in the cholesterol-binding domain of APP or APP null alleles, we found that while CE also regulate Aß secretion, the effects of CE on Tau and Aß are mediated by independent pathways. Efficacy and toxicity screening in iPSC-derived astrocytes and neurons showed that allosteric activation of CYP46A1 lowers CE specifically in neurons and is well tolerated by astrocytes. These data reveal that CE independently regulate Tau and Aß and identify a druggable CYP46A1-CE-Tau axis in AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Colesterol/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/patologia , Animais , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL
7.
Stem Cell Reports ; 10(3): 1046-1058, 2018 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-29503090

RESUMO

Developing effective therapeutics for complex diseases such as late-onset, sporadic Alzheimer's disease (SAD) is difficult due to genetic and environmental heterogeneity in the human population and the limitations of existing animal models. Here, we used hiPSC-derived neurons to test a compound that stabilizes the retromer, a highly conserved multiprotein assembly that plays a pivotal role in trafficking molecules through the endosomal network. Using this human-specific system, we have confirmed previous data generated in murine models and show that retromer stabilization has a potentially beneficial effect on amyloid beta generation from human stem cell-derived neurons. We further demonstrate that manipulation of retromer complex levels within neurons affects pathogenic TAU phosphorylation in an amyloid-independent manner. Taken together, our work demonstrates that retromer stabilization is a promising candidate for therapeutic development in AD and highlights the advantages of testing novel compounds in a human-specific, neuronal system.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Fosforilação/fisiologia , Proteínas tau/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Células Cultivadas , Endossomos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Neurônios/metabolismo , Transporte Proteico/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa